Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Proc Natl Acad Sci U S A ; 117(33): 19854-19865, 2020 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-32759214

RESUMO

The blood-retina barrier and blood-brain barrier (BRB/BBB) are selective and semipermeable and are critical for supporting and protecting central nervous system (CNS)-resident cells. Endothelial cells (ECs) within the BRB/BBB are tightly coupled, express high levels of Claudin-5 (CLDN5), a junctional protein that stabilizes ECs, and are important for proper neuronal function. To identify novel CLDN5 regulators (and ultimately EC stabilizers), we generated a CLDN5-P2A-GFP stable cell line from human pluripotent stem cells (hPSCs), directed their differentiation to ECs (CLDN5-GFP hPSC-ECs), and performed flow cytometry-based chemogenomic library screening to measure GFP expression as a surrogate reporter of barrier integrity. Using this approach, we identified 62 unique compounds that activated CLDN5-GFP. Among them were TGF-ß pathway inhibitors, including RepSox. When applied to hPSC-ECs, primary brain ECs, and retinal ECs, RepSox strongly elevated barrier resistance (transendothelial electrical resistance), reduced paracellular permeability (fluorescein isothiocyanate-dextran), and prevented vascular endothelial growth factor A (VEGFA)-induced barrier breakdown in vitro. RepSox also altered vascular patterning in the mouse retina during development when delivered exogenously. To determine the mechanism of action of RepSox, we performed kinome-, transcriptome-, and proteome-profiling and discovered that RepSox inhibited TGF-ß, VEGFA, and inflammatory gene networks. In addition, RepSox not only activated vascular-stabilizing and barrier-establishing Notch and Wnt pathways, but also induced expression of important tight junctions and transporters. Taken together, our data suggest that inhibiting multiple pathways by selected individual small molecules, such as RepSox, may be an effective strategy for the development of better BRB/BBB models and novel EC barrier-inducing therapeutics.


Assuntos
Células Endoteliais/efeitos dos fármacos , Células-Tronco Pluripotentes/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/farmacologia , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Barreira Hematorretiniana/efeitos dos fármacos , Barreira Hematorretiniana/metabolismo , Diferenciação Celular , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Claudina-5/genética , Claudina-5/metabolismo , Avaliação Pré-Clínica de Medicamentos , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Edição de Genes , Genoma , Humanos , Camundongos , Camundongos Knockout , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Pirazóis/farmacologia , Piridinas/farmacologia , Junções Íntimas/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
2.
Pharm Res ; 35(9): 169, 2018 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-29951887

RESUMO

PURPOSE: Administration of therapeutic monoclonal antibodies (mAbs) is frequently accompanied by severe first infusion reactions (FIR). The mechanism driving FIR is still unclear. This study aimed to investigate the cellular and molecular mechanisms causing FIR in humanized mouse models and their potential for evaluating FIR risk in patients. METHODS: Mice humanized for Fc gamma receptors (FcγRs) were generated by recombination-mediated genomic replacement. Body temperature, cytokine release and reactive oxygen species (ROS) were measured to assess FIR to mAbs. RESULTS: Infusion of human mAb specific for mouse transferrin receptor (HamTfR) into FcγR-humanized mice, produced marked transient hypothermia accompanied by an increase in inflammatory cytokines KC and MIP-2, and ROS. FIR were dependent on administration route and Fc-triggered effector functions mediated by neutrophils. Human neutrophils also induced FIR in wild type mice infused with HamTfR. Specific knock-in mice demonstrated that human FcγRIIIb on neutrophils was both necessary and sufficient to cause FIR. FcγRIIIb-mediated FIR was abolished by depleting neutrophils or blocking FcγRIIIb with CD11b antibodies. CONCLUSIONS: Human FcγRIIIb and neutrophils are primarily responsible for triggering FIR. Clinical strategies to prevent FIR in patients should focus on this pathway and may include transient depletion of neutrophils or blocking FcγRIIIb with specific mAbs.


Assuntos
Anticorpos Monoclonais/efeitos adversos , Hipotermia/induzido quimicamente , Inflamação/induzido quimicamente , Neutrófilos/imunologia , Receptores de IgG/imunologia , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/imunologia , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/imunologia , Humanos , Hipotermia/imunologia , Inflamação/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neutrófilos/efeitos dos fármacos , Receptores de IgG/genética , Receptores da Transferrina/imunologia
3.
Proc Natl Acad Sci U S A ; 108(20): 8485-90, 2011 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-21525407

RESUMO

The trace amine-associated receptor 1 (TAAR1), activated by endogenous metabolites of amino acids like the trace amines p-tyramine and ß-phenylethylamine, has proven to be an important modulator of the dopaminergic system and is considered a promising target for the treatment of neuropsychiatric disorders. To decipher the brain functions of TAAR1, a selective TAAR1 agonist, RO5166017, was engineered. RO5166017 showed high affinity and potent functional activity at mouse, rat, cynomolgus monkey, and human TAAR1 stably expressed in HEK293 cells as well as high selectivity vs. other targets. In mouse brain slices, RO5166017 inhibited the firing frequency of dopaminergic and serotonergic neurons in regions where Taar1 is expressed (i.e., the ventral tegmental area and dorsal raphe nucleus, respectively). In contrast, RO5166017 did not change the firing frequency of noradrenergic neurons in the locus coeruleus, an area devoid of Taar1 expression. Furthermore, modulation of TAAR1 activity altered the desensitization rate and agonist potency at 5-HT(1A) receptors in the dorsal raphe, suggesting that TAAR1 modulates not only dopaminergic but also serotonergic neurotransmission. In WT but not Taar1(-/-) mice, RO5166017 prevented stress-induced hyperthermia and blocked dopamine-dependent hyperlocomotion in cocaine-treated and dopamine transporter knockout mice as well as hyperactivity induced by an NMDA antagonist. These results tie TAAR1 to the control of monoamine-driven behaviors and suggest anxiolytic- and antipsychotic-like properties for agonists such as RO5166017, opening treatment opportunities for psychiatric disorders.


Assuntos
Monoaminas Biogênicas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transmissão Sináptica/fisiologia , Animais , Benzodioxóis/farmacologia , Dopamina/metabolismo , Glutamina/metabolismo , Células HEK293 , Humanos , Transtornos Mentais , Camundongos , Fenilpropionatos/farmacologia , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/deficiência
4.
Sci Rep ; 10(1): 10542, 2020 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-32601281

RESUMO

Naïve human pluripotent stem cells (hPSC) resemble the embryonic epiblast at an earlier time-point in development than conventional, 'primed' hPSC. We present a comprehensive miRNA profiling of naïve-to-primed transition in hPSC, a process recapitulating aspects of early in vivo embryogenesis. We identify miR-143-3p and miR-22-3p as markers of the naïve state and miR-363-5p, several members of the miR-17 family, miR-302 family as primed markers. We uncover that miR-371-373 are highly expressed in naïve hPSC. MiR-371-373 are the human homologs of the mouse miR-290 family, which are the most highly expressed miRNAs in naïve mouse PSC. This aligns with the consensus that naïve hPSC resemble mouse naive PSC, showing that the absence of miR-371-373 in conventional hPSC is due to cell state rather than a species difference.


Assuntos
Perfilação da Expressão Gênica , Células-Tronco Pluripotentes Induzidas/metabolismo , MicroRNAs/metabolismo , Células-Tronco Pluripotentes/metabolismo , Humanos , MicroRNAs/genética
5.
Front Pharmacol ; 9: 924, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30158871

RESUMO

Individuals of many species rely on odors to communicate, find breeding partners, locate resources and sense dangers. In vertebrates, odorants are detected by chemosensory receptors of the olfactory system. One class of these receptors, the trace amine-associated receptors (TAARs), was recently suggested to mediate male sexual interest and mate choice. Here we tested this hypothesis in mice by generating a cluster deletion mouse (Taar2-9-/-) lacking all TAARs expressed in the olfactory epithelium, and evaluating transduction pathways from odorants to TAARs, neural activity and behaviors reflecting sexual interest. We found that a urinary volatile amine, isobutylamine (IBA), was a potent ligand for TAAR3 (but not TAAR1, 4, 5, and 6). When males were exposed to IBA, brain regions associated with sexual behaviors were less active in Taar2-9-/- than in wild type males. Accordingly, Taar2-9-/- males spent less time sniffing both the urine of females and pure IBA than wild type males. This is the first demonstration of a comprehensive transduction pathway linking odorants to TAARs and male sexual interest. Interestingly, the concentration of IBA in female urine varied across the estrus cycle with a peak during estrus. This variation in IBA concentration may represent a simple olfactory cue for males to recognize receptive females. Our results are consistent with the hypothesis that IBA and TAARs play an important role in the recognition of breeding partners and mate choice.

6.
Curr Biol ; 12(8): 661-6, 2002 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-11967154

RESUMO

Inactivation of Cyclin E-Cdk2 is essential for a timely arrest of the epidermal cell proliferation program during Drosophila embryogenesis. E-type cyclin-cdk complexes are thought to be activated by D-types titrating away inhibitors and inducing cyclin E transcription by activating E2F transcription factors via Rb phosphorylation. Therefore, we have analyzed whether the developmentally controlled inactivation of Cyclin E-Cdk2 required for the epidermal cell proliferation arrest occurs as a consequence of Cyclin D-Cdk4 inactivation. However, preventing Cyclin D-Cdk4 inactivation by overexpression has a minimal effect on Cyclin E expression and does not interfere with the initial G1 arrest, while it readily induces the E2F target RnrS in arresting epidermal cells. Prolonged Cyclin D-Cdk4 overexpression eventually interferes with maintenance of quiescence in some cells. Moreover, in Cdk4 mutant embryos, some RnrS expression is still induced by Cyclin E overexpression, and endogenous Cyclin E expression as well as cell cycle progression is not affected, except for late aspects of the endoreduplication program. These findings argue against the proposed necessity of complete Rb inactivation by sequential phosphorylation by D- and E-type cyclin-cdk complexes. They demonstrate that Cyclin D-Cdk4 does not function as the master regulator of the embryonic cell proliferation program.


Assuntos
Proteínas de Ciclo Celular , Quinases Ciclina-Dependentes/metabolismo , Ciclinas/metabolismo , Proteínas de Ligação a DNA , Drosophila/citologia , Drosophila/embriologia , Proteínas Proto-Oncogênicas , Animais , Divisão Celular , Ciclina D , Ciclina E/genética , Ciclina E/metabolismo , Quinase 4 Dependente de Ciclina , Quinases Ciclina-Dependentes/genética , Ciclinas/genética , Drosophila/genética , Drosophila/metabolismo , Proteínas de Drosophila , Fatores de Transcrição E2F , Expressão Gênica , Fase S , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
7.
Cell Rep ; 15(1): 86-95, 2016 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-27052171

RESUMO

Hyperfunction of the mTORC1 pathway has been associated with idiopathic and syndromic forms of autism spectrum disorder (ASD), including tuberous sclerosis, caused by loss of either TSC1 or TSC2. It remains largely unknown how developmental processes and biochemical signaling affected by mTORC1 dysregulation contribute to human neuronal dysfunction. Here, we have characterized multiple stages of neurogenesis and synapse formation in human neurons derived from TSC2-deleted pluripotent stem cells. Homozygous TSC2 deletion causes severe developmental abnormalities that recapitulate pathological hallmarks of cortical malformations in patients. Both TSC2(+/-) and TSC2(-/-) neurons display altered synaptic transmission paralleled by molecular changes in pathways associated with autism, suggesting the convergence of pathological mechanisms in ASD. Pharmacological inhibition of mTORC1 corrects developmental abnormalities and synaptic dysfunction during independent developmental stages. Our results uncouple stage-specific roles of mTORC1 in human neuronal development and contribute to a better understanding of the onset of neuronal pathophysiology in tuberous sclerosis.


Assuntos
Complexos Multiproteicos/antagonistas & inibidores , Células-Tronco Neurais/metabolismo , Neurogênese , Sinapses/metabolismo , Serina-Treonina Quinases TOR/antagonistas & inibidores , Esclerose Tuberosa/metabolismo , Linhagem Celular , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina , Complexos Multiproteicos/metabolismo , Células-Tronco Neurais/citologia , Células-Tronco Neurais/fisiologia , Sinapses/fisiologia , Transmissão Sináptica , Serina-Treonina Quinases TOR/metabolismo , Esclerose Tuberosa/genética , Proteína 2 do Complexo Esclerose Tuberosa , Proteínas Supressoras de Tumor/genética
8.
Genetics ; 168(2): 867-75, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15514060

RESUMO

The three mammalian D-type cyclins are thought to promote progression through the G1 phase of the cell cycle as regulatory subunits of cyclin-dependent kinase 4 and 6. In addition, they have been proposed to control the activity of various transcription factors without a partner kinase. Here we describe phenotypic consequences of null mutations in Cyclin D, the single D-type cyclin gene in Drosophila. As previously observed with null mutations in the single Drosophila Cdk4 gene, these mutations do not primarily affect progression through the G1 phase. Moreover, the apparently indistinguishable phenotypes of double (CycD and Cdk4) and single mutants (CycD or Cdk4) argue against major independent functions of Cyclin D and Cdk4. The reduced cellular and organismal growth rates observed in both mutants indicate that Cyclin D-Cdk4 acts as a growth driver.


Assuntos
Divisão Celular/fisiologia , Quinases Ciclina-Dependentes/metabolismo , Ciclinas/fisiologia , Drosophila melanogaster/crescimento & desenvolvimento , Fase G1 , Mutação/genética , Proteínas Proto-Oncogênicas/metabolismo , Animais , Animais Geneticamente Modificados , Apoptose , Peso Corporal , Bromodesoxiuridina , Ciclina D , Quinase 4 Dependente de Ciclina , Proteínas de Drosophila , Drosophila melanogaster/enzimologia , Drosophila melanogaster/genética , Feminino , Masculino , Fenótipo , Asas de Animais/citologia , Asas de Animais/metabolismo
9.
Eur Neuropsychopharmacol ; 25(11): 2049-61, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26372541

RESUMO

Trace amine-associated receptor 1 (TAAR1) activation by selective endogenous agonists modulates dopaminergic neurotransmission. This results in antipsychotic-like behavior in vivo which might be initiated by an interaction of TAAR1 and dopamine D2L receptor (D2R). Here we analyzed the functional link between TAAR1 and D2R using highly potent and selective TAAR1 agonists, and newly generated tools such as TAAR1 knock-out and TAAR1 overexpressing rats as well as specific anti-rat TAAR1 antibodies. We provide data from co-immunoprecipitation experiments supporting a functional interaction of the two receptors in heterologous cells and in brain tissue. Interaction of TAAR1 with D2R altered the subcellular localization of TAAR1 and increased D2R agonist binding affinity. Using specific ß-arrestin 2 (ßArr2) complementation assays we show that the interaction of TAAR1 with D2R reduced ßArr2 recruitment to D2R. In addition, we report that besides Gαs-protein signaling TAAR1 also signals via ßArr2. In the presence of D2R, cAMP signaling of TAAR1 was reduced while its ßArr2 signaling was enhanced, resulting in reduced GSK3ß activation. These results demonstrate that ßArr2 signaling may be an important pathway for TAAR1 function and that the activation of the TAAR1-D2R complex negatively modulates GSK3ß signaling. Given that patients with schizophrenia or bipolar disorder show increased GSK3ß signaling, such a reduction of GSK3ß signaling triggered by the interaction of D2R with activated TAAR1 further supports TAAR1 as a target for the treatment of psychiatric disorders.


Assuntos
Arrestinas/metabolismo , Encéfalo/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Receptores de Dopamina D2/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animais , Células CHO , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Cricetulus , AMP Cíclico/metabolismo , Técnicas de Inativação de Genes , Glicogênio Sintase Quinase 3 beta , Células HEK293 , Humanos , Ratos Transgênicos , Ratos Wistar , Receptores de Dopamina D2/agonistas , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/genética , beta-Arrestina 2 , beta-Arrestinas
10.
Nat Cell Biol ; 17(1): 57-67, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25487280

RESUMO

The rising incidence of obesity and related disorders such as diabetes and heart disease has focused considerable attention on the discovery of new therapeutics. One promising approach has been to increase the number or activity of brown-like adipocytes in white adipose depots, as this has been shown to prevent diet-induced obesity and reduce the incidence and severity of type 2 diabetes. Thus, the conversion of fat-storing cells into metabolically active thermogenic cells has become an appealing therapeutic strategy to combat obesity. Here, we report a screening platform for the identification of small molecules capable of promoting a white-to-brown metabolic conversion in human adipocytes. We identified two inhibitors of Janus kinase (JAK) activity with no precedent in adipose tissue biology that stably confer brown-like metabolic activity to white adipocytes. Importantly, these metabolically converted adipocytes exhibit elevated UCP1 expression and increased mitochondrial activity. We further found that repression of interferon signalling and activation of hedgehog signalling in JAK-inactivated adipocytes contributes to the metabolic conversion observed in these cells. Our findings highlight a previously unknown role for the JAK-STAT pathway in the control of adipocyte function and establish a platform to identify compounds for the treatment of obesity.


Assuntos
Adipócitos Marrons/citologia , Adipócitos Brancos/citologia , Janus Quinase 3/antagonistas & inibidores , Oxazinas/farmacologia , Piperidinas/farmacologia , Piridinas/farmacologia , Pirimidinas/farmacologia , Pirróis/farmacologia , Animais , Proteína Morfogenética Óssea 7 , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Perfilação da Expressão Gênica , Proteínas Hedgehog/metabolismo , Humanos , Interferon gama/biossíntese , Interferon gama/farmacologia , Canais Iônicos/biossíntese , Janus Quinase 1/antagonistas & inibidores , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/metabolismo , Proteínas Mitocondriais/biossíntese , Obesidade/prevenção & controle , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Fator de Transcrição STAT1/antagonistas & inibidores , Fator de Transcrição STAT3/antagonistas & inibidores , Fator de Necrose Tumoral alfa/farmacologia , Proteína Desacopladora 1 , Alcaloides de Veratrum/farmacologia
11.
Neuropsychopharmacology ; 37(12): 2580-92, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22763617

RESUMO

Trace amines (TAs) such as ß-phenylethylamine, p-tyramine, or tryptamine are biogenic amines found in the brain at low concentrations that have been implicated in various neuropsychiatric disorders like schizophrenia, depression, or attention deficit hyperactivity disorder. TAs are ligands for the recently identified trace amine-associated receptor 1 (TAAR1), an important modulator of monoamine neurotransmission. Here, we sought to investigate the consequences of TAAR1 hypersignaling by generating a transgenic mouse line overexpressing Taar1 specifically in neurons. Taar1 transgenic mice did not show overt behavioral abnormalities under baseline conditions, despite augmented extracellular levels of dopamine and noradrenaline in the accumbens nucleus (Acb) and of serotonin in the medial prefrontal cortex. In vitro, this was correlated with an elevated spontaneous firing rate of monoaminergic neurons in the ventral tegmental area, dorsal raphe nucleus, and locus coeruleus as the result of ectopic TAAR1 expression. Furthermore, Taar1 transgenic mice were hyposensitive to the psychostimulant effects of amphetamine, as it produced only a weak locomotor activation and failed to alter catecholamine release in the Acb. Attenuating TAAR1 activity with the selective partial agonist RO5073012 restored the stimulating effects of amphetamine on locomotion. Overall, these data show that Taar1 brain overexpression causes hyposensitivity to amphetamine and alterations of monoaminergic neurotransmission. These observations confirm the modulatory role of TAAR1 on monoamine activity and suggest that in vivo the receptor is either constitutively active and/or tonically activated by ambient levels of endogenous agonist(s).


Assuntos
Anfetamina/farmacologia , Monoaminas Biogênicas/fisiologia , Química Encefálica/fisiologia , Estimulantes do Sistema Nervoso Central/farmacologia , Receptores Acoplados a Proteínas G/biossíntese , Receptores Acoplados a Proteínas G/fisiologia , Transmissão Sináptica/fisiologia , Compostos de Anilina/farmacologia , Animais , Comportamento Animal/efeitos dos fármacos , Neurônios Dopaminérgicos/efeitos dos fármacos , Fenômenos Eletrofisiológicos , Imidazóis/farmacologia , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos C57BL , Microdiálise , Atividade Motora/efeitos dos fármacos , Atividade Motora/fisiologia , Técnicas de Patch-Clamp , Fenótipo , Área Tegmentar Ventral/efeitos dos fármacos , Área Tegmentar Ventral/fisiologia , Ácido gama-Aminobutírico/fisiologia
12.
J Immunol ; 179(9): 5644-8, 2007 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-17947634

RESUMO

Sphingosine kinase (Sphk) phosphorylates sphingosine into sphingosine-1-phosphate (S1P), but its recently identified isoform Sphk2 has been suggested to have distinct subcellular localization and substrate specificity. We demonstrate here that, surprisingly, Sphk2(-/-) CD4(+) T cells exhibit a hyperactivated phenotype with significantly enhanced proliferation and cytokine secretion in response to IL-2 as well as reduced sensitivity to regulatory T cell-mediated suppression in vitro, apparently independent of effects upon S1P. Such findings appear to reflect a requirement for Sphk2 to suppress IL-2 signaling because, in Sphk2(-/-) CD4(+) T cells, IL-2 induced abnormally accentuated STAT5 phosphorylation and small interfering RNA knockdown of STAT5 abrogated their hyperactive phenotype. This pathway physiologically modulates autoinflammatory responses, because Sphk2(-/-) T cells induced more rapid and robust inflammatory bowel disease in scid recipients. Thus, Sphk2 regulates IL-2 pathways in T cells, and the modulation of Sphk2 activity may be of therapeutic utility in inflammatory and/or infectious diseases.


Assuntos
Autoimunidade/imunologia , Interleucina-2/imunologia , Interleucina-2/metabolismo , Mucosa Intestinal/metabolismo , Intestinos/imunologia , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Transdução de Sinais/imunologia , Animais , Doenças Inflamatórias Intestinais/enzimologia , Doenças Inflamatórias Intestinais/imunologia , Doenças Inflamatórias Intestinais/patologia , Lisofosfolipídeos/metabolismo , Camundongos , Camundongos Knockout , Fenótipo , Fosfotransferases (Aceptor do Grupo Álcool)/deficiência , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Linfócitos T/enzimologia , Linfócitos T/imunologia
13.
Development ; 129(2): 319-28, 2002 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11807025

RESUMO

dacapo encodes a CIP/KIP-type inhibitor of Cyclin E/Cdk2 complexes in Drosophila melanogaster. In the embryonic epidermis, dacapo expression starts during G2 of the final division cycle and is required for the arrest of cell cycle progression in G1 after the final mitosis. The onset of dacapo transcription is the earliest event known to be required for the epidermal cell proliferation arrest. To advance our understanding of the regulatory mechanisms that terminate cell proliferation at the appropriate stage, we have analyzed the control of dacapo transcription. We show that dacapo transcription is not coupled to cell cycle progression. It is not affected in mutants where proliferation is arrested either too early or too late. Moreover, upregulation of dacapo expression is not an obligatory event of the cell cycle exit process. During early development of the central nervous system, we cannot detect p27Dacapo during the final division cycle of ganglion mother cells, while it is expressed at later stages. The control of dacapo expression therefore varies in different stages and tissues. The dacapo regulatory region includes many independent cis-regulatory elements. The elements that control epidermal expression integrate developmental cues that time the arrest of cell proliferation.


Assuntos
Ciclo Celular/fisiologia , Proteínas de Drosophila , Drosophila melanogaster/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Proteínas Nucleares/genética , Proteínas Tirosina Fosfatases , Sequências Reguladoras de Ácido Nucleico , Transcrição Gênica , Animais , Padronização Corporal/genética , Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Inibidor de Quinase Dependente de Ciclina p27 , Quinases Ciclina-Dependentes/antagonistas & inibidores , Quinases Ciclina-Dependentes/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Inibidores Enzimáticos/metabolismo , Genes Reporter , Hibridização In Situ , Proteínas de Insetos/genética , Proteínas de Insetos/metabolismo , Proteínas Nucleares/metabolismo , Fosfoproteínas Fosfatases/genética , Fosfoproteínas Fosfatases/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Transgenes , Proteínas Supressoras de Tumor/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA